Superoxide anion is implicated in the regulation of PP2A b56 mediated dephosphorylation of bcl 2

230 272 0
Superoxide anion is implicated in the regulation of PP2A b56  mediated dephosphorylation of bcl 2

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

SUPEROXIDE ANION IS IMPLICATED IN THE REGULATION OF PP2AB56δ-MEDIATED DEPHOSPHORYLATION OF BCL-2 IVAN LOW CHERH CHIET [BSc. (Hons.), NUS] A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS GRADUATE SCHOOL FOR INTEGRATIVE SCIENCES AND ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2012 Declaration I hereby declare that this thesis is my original work and it has been written by me in its entirety. I have duly acknowledged all the sources of information which have been used in the thesis. This thesis has also not been submitted for any degree in any university previously. ________________________ Ivan Low Cherh Chiet 07 Nov 2012 ACKNOWLEDGEMENTS Four years of fulfilling and enriching experience; of joy, excitement and laughter; of sorrows, disappointments and despair. It was unique, but none possible without the company and contributions of these people. My utmost gratitude goes to them. To Prof Shazib Pervaiz – a supervisor, a boss and also a friend. It is your relentless guidance, enthusiasm and support that have brought me thus far. The scientific and intellectual brain teasers imparted throughout the course of my PhD is amongst the biggest drivers underlying what I have achieved today. I sincerely thank you, not only for the intellectual contributions, but also the countless opportunities that you have created for me. And not to forget, the wonderful experience we have shared over numerous conference trips as well as the time spent running together along the roads, bahut shukriya. To my thesis advisory committee (TAC) members, Prof David Marc Virshup and Assoc. Prof Marie-Veronique Clement. Thank you for the ever critical and constructive inputs, as well as the reagents and experimental tools that you have generously shared with me. Merci. To my lab mates, Tini and Maj, for tolerating my last minute orders and my irritating pesters over urgent orders, terima kasih. To Jay, for being the lab’s mother since I have first stepped into the lab as an innocent honours student, dhanyavad. To Stephen, for taking all my scoldings and naggings positively, even though they often fall on deaf ears, toh cheh. To Pat, Serena, Angeline, Carolyn, Zhou Ting and the rest of my ex- and current lab mates, thanks for sharing the wonderful working hours as well as the limited lab space and equipment with me. Thank you all! i To my hall and running friends, especially Kuan Thye, Teik Zhen, Aaron, Roonz, Eugene Tan, twin bro Lee, Huat and Alan. Thank you for spicing up my life other than lab, research, studies and work. To my parents and my younger sister, thank you for the guidance, patience, concern and support since my birth, and throughout the entire period when I am in Singapore. Mum and dad, the baby that had once cried off your sleeping hours as well as the boy that had frequently gotten your blood pressure raised hopes that he could you proud here. Last but not least, my wife, Hui Ting, words cannot describe the gratitude I have for all the support, love and care that you have rendered unconditionally. It is you that kept me going when I am down, defeated, and injured, but I am never out all because of you. Thanks for sharing my joy and happiness, my sorrows and hardships. Through thick and thin, I love you. ii TABLE OF CONTENTS ACKNOWLEDGEMENTS . i TABLE OF CONTENTS iii SUMMARY . ix LIST OF TABLES xi LIST OF FIGURES xi LIST OF ABBREVIATIONS . xv INTRODUCTION . 1 Reactive Oxygen Species – An overview 1.1 Superoxide anion (O2-) 1.2 Hydrogen Peroxide (H2O2) . 1.3 Nitric Oxide (NO) . 1.4 Peroxynitrite (ONOO-) 10 Cellular Antioxidant Defences . 12 2.1 Superoxide Dismutases . 12 2.2 Catalase . 13 2.3 Gluthathione Peroxidases 14 2.4 Peroxiredoxins and Thioredoxins . 15 Apoptotic Cell Death 16 3.1 Caspases – the mastermind of apoptosis . 17 3.2 Extrinsic pathway 18 3.3 Intrinsic pathway . 19 3.4 The anti-apoptotic B Cell Lymphoma-2 protein . 22 ROS and the apoptotic cell death machinery . 27 iii 4.1 ROS as biphasic regulators of apoptosis . 28 4.2 Oncogene-mediated pro-oxidant state – Bcl-2 as a prime example 31 Protein Phosphatase 2A 32 5.1 The tumour suppressive role of PP2A 34 5.2 PP2A and the apoptotic cell death machinery . 36 Aims and Objectives . 38 MATERIALS AND METHODS 39 Cell lines and cell culture . 39 Reagents and chemicals 39 Antibodies . 40 Plasmids and SiRNAs . 41 Calcium phosphate-based transfection of adherent cells 42 Electroporation-based transfection of Jurkat cells . 43 Determination of protein concentration 44 Western blot analysis 44 Co-immunoprecipitation (co-IP) assay . 45 10 Mitochondrial-cytoplasmic fractionation . 46 11 Immunofluorescence confocal microscopy 47 12 MTT cell viability assay . 49 13 Crystal violet cell viability assay 49 14 Caspase activity assay . 50 15 Measurement of cellular O2- via Lucigenin chemiluminescence assay 51 16 Flow cytometry analysis of intracellular NO 51 17 Measurement of PP2A activity (serine/threonine phosphatase assay) . 52 18 Construction of Y289F-B56δ mutant via site-directed mutagenesis 54 iv 19 Extraction and purification of primary lymphoma tumours . 56 20 Statistical analysis . 57 RESULTS . 58 O2- promotes tumour chemoresistance by inducing Serine70 Bcl-2 phosphorylation . 58 1.1 Diethyldithiocarbamate (DDC) induced a parallel increase in both intracellular O2- and S70 pBcl-2 . 58 1.2 Scavenging of O2- abrogated DDC-induced S70 bcl-2 phosphorylation 61 1.3 SiRNA-mediated downregulation of SOD1 mimicked the effect of DDC . . 63 1.4 Treatment of bovine SOD1 led to a reduction in S70 pBcl-2 . 65 1.5 Pre-treatment of DDC promoted chemoresistance in tumour cells 67 1.6 Pre-treatment of DDC abated etoposide- and doxorubicin-induced activation of caspase-9 and caspase-3 69 1.7 Silencing of SOD1 protected against doxorubicin- and etoposide-induced cell death 71 1.8 S70 phosphorylation of Bcl-2 is required for the death-inhibitory activity of DDC . 72 1.9 S70 phosphorylation of Bcl-2 did not enhance its ability to sequester the pro-apoptotic protein, BAK . 75 O2- induced S70 pBcl-2 upregulation by inactivating the Bcl-2 phosphatase, PP2A 77 2.1 O2--induced S70 pBcl-2 upregulation is not a function of MAP kinases activation 77 2.2 O2- generated by pharmacological inhibition or genetic knockdown of SOD1 triggered a drop in PP2A activity 80 2.3 O2- inhibited the interaction between Bcl-2 and the catalytic subunit of PP2A . 82 2.4 Co-localization of PP2A-C and Bcl-2 was inhibited in cells with an augmented O2- level . 85 v 2.5 Mitochondrial translocation of PP2A-C was inhibited in tumour cells with heightened intracellular O2- level . 90 O2- augmented the level of S70 pBcl-2 by inhibiting the holoenzyme assembly of B56δ-containing PP2A (PP2AB56δ) . 94 3.1 The B56δ regulatory subunit of PP2A is responsible for the substrate recognition of Bcl-2 in Jurkat and MDA-231 cells 94 3.2 O2- inhibited the binding of the AC catalytic core, but not B56δ, to Bcl-2 . . 97 3.3 DDC treatment did not affect the co-localization of B56δ and Bcl-2 . 99 3.4 SiRNA-mediated knockdown of SOD1 inhibited the interaction of Bcl-2 with PP2A-C but not with B56δ 100 3.5 O2- inhibited mitochondrial localization of PP2A-AC catalytic core . 102 ONOO- as the missing link between O2- and the inhibition of PP2A 104 4.1 DDC treatment of tumour cells was accompanied by a drop in intracellular nitric oxide (NO) level . 104 4.2 ONOO- is necessary for DDC-induced S70 Bcl-2 phosphorylation . 106 4.3 Low doses of ONOO- mirrored the effects of O2- in promoting S70 pBcl-2 and chemoresistance . 107 4.4 ONOO- treatment mimicked the inhibitory effect of SOD1 downregulation on the interaction between B56δ and the AC catalytic core 110 4.5 O2- inhibited mitochondrial localization of PP2A-AC catalytic core via the production of ONOO- . 112 O2- inhibited PP2AB56δ holoenzyme assembly via ONOO--mediated nitration of B56δ at tyrosine-289 (Y289) . 113 5.1 Bioinformatics analysis revealed a conserved tyrosine residue on PP2AB56 regulatory subunits that is prone to nitrative modification . 113 5.2 DDC treatment augmented the level of 3-nitrotyrosine (3-NT) in HAB56α and HA-B56δ 117 5.3 3-NT level was elevated in endogenous B56δ upon DDC and ONOOtreatment . 119 vi 5.4 SOD1 knockdown induced B56δ tyrosine nitration and inhibited B56δmediated recruitment of PP2A-C to Bcl-2 . 121 5.5 Inhibition of NOX by diphenyliodium (DPI) abrogated the inhibitory effect of SOD1 knockdown on PP2AB56δ . 124 5.6 Scavenging of O2- via bSOD1 treatment potentiated PP2AB56δ mediated dephosphorylation of Bcl-2 at S70 . 126 5.7 DDC-induced O2- production also stimulated the phosphorylation of tau, but not pRb, and Raf 128 5.8 Nitration status of B56δY289 is positively correlated with the level of S70 pBcl-2 in Jurkat cells 130 5.9 Site-directed mutagenesis studies confirmed the regulatory role of Y289 B56δ nitration in O2--induced upregulation of S70 pBcl-2 133 5.10 Y289F-B56δ transfection restored the interaction between B56δ and PP2A-C in DDC-treated Jurkat cells 136 5.11 Y289F-B56δ transfection restored the co-localization status of Bcl-2 and PP2A-C in DDC-treated Jurkat cells 139 5.12 Y289F-B56δ transfection abolished the death-inhibitory effect of DDC in Jurkat T-leukemic cells 144 Clinical relevance of SOD1 downregulation in primary lymphoma biopsy samples . 146 6.1 An inverse relationship exists between SOD1 and S70 pBcl-2 in primary lymphoma cells 146 6.2 Primary lymphoma tumours expressing high level of S70 pBcl-2 harboured B56δ that are tyrosine nitrated 149 DISCUSSION . 151 ROS as intricate regulators of cell death 151 1.1 S70 pBcl-2 as a key player in O2--mediated chemoresistance 151 1.2 O2- and ONOO- – Killers or protectors? 152 1.3 SOD1 – are they tumour suppressors? 154 S70 pBcl-2 – How it is regulated, and how it regulates . 159 vii 2.1 Redox regulation of Bcl-2 phosphorylation 159 2.2 Potential source(s) of O2- and NO responsible for the induction of S70 pBcl-2 . 160 2.3 The death-regulatory role of S70 pBcl-2 165 A new facet in the anti-apoptotic activity of Bcl-2 170 Inhibitory tyrosine nitration of B56 regulatory isoforms – potential implication(s) beyond the activation of Bcl-2 . 172 4.1 Redox regulation of PP2AB56δ – implications on phospho-tau 172 4.2 B56 regulatory subunits as strategic targets for O2--mediated carcinogenesis 173 4.3 The fate of B56δ-dissociated PP2A-C 175 O2--mediated inhibition of PP2A – its relevance in a clinical setting 177 5.1 Is PP2A a druggable target for anti-cancer therapy? . 177 5.2 Activating specific PP2A complexes as a novel chemotherapeutic strategy . 180 5.3 Targeting intracellular O2- as a novel therapeutic strategy against Bcl-2induced chemoresistance 182 5.4 SOD1, S70 pBcl-2 and B56δY289 nitration (nitro-B56δY289) – a potential prognostic signature for better cancer management and care 183 Conclusion . 186 REFERENCES 189 APPENDICES 209 List of Publications 209 Conference Papers . 209 viii 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. Meister, A., Glutathione metabolism and its selective modification. J Biol Chem, 1988. 263(33): p. 17205-8. Schafer, F.Q. and G.R. Buettner, Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med, 2001. 30(11): p. 1191-212. Flohe, L., H. Budde, and B. Hofmann, Peroxiredoxins in antioxidant defense and redox regulation. Biofactors, 2003. 19(1-2): p. 3-10. Hofmann, B., H.J. Hecht, and L. Flohe, Peroxiredoxins. Biol Chem, 2002. 383(3-4): p. 347-64. Rhee, S.G., et al., Peroxiredoxin, a novel family of peroxidases. IUBMB Life, 2001. 52(1-2): p. 35-41. Seo, M.S., et al., Identification of a new type of mammalian peroxiredoxin that forms an intramolecular disulfide as a reaction intermediate. J Biol Chem, 2000. 275(27): p. 20346-54. Rhee, S.G., H.Z. Chae, and K. Kim, Peroxiredoxins: a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic Biol Med, 2005. 38(12): p. 1543-52. Holmgren, A. and M. Bjornstedt, Thioredoxin and thioredoxin reductase. Methods Enzymol, 1995. 252: p. 199-208. Chae, H.Z., S.J. Chung, and S.G. Rhee, Thioredoxin-dependent peroxide reductase from yeast. J Biol Chem, 1994. 269(44): p. 27670-8. Kerr, J.F., A.H. Wyllie, and A.R. Currie, Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer, 1972. 26(4): p. 239-57. Wyllie, A.H., J.F. Kerr, and A.R. Currie, Cell death: the significance of apoptosis. Int Rev Cytol, 1980. 68: p. 251-306. Jacobson, M.D., M. Weil, and M.C. Raff, Programmed cell death in animal development. Cell, 1997. 88(3): p. 347-54. Osborne, B.A., Apoptosis and the maintenance of homoeostasis in the immune system. Curr Opin Immunol, 1996. 8(2): p. 245-54. Hanahan, D. and R.A. Weinberg, The hallmarks of cancer. Cell, 2000. 100(1): p. 57-70. Hanahan, D. and R.A. Weinberg, Hallmarks of cancer: the next generation. Cell, 2011. 144(5): p. 646-74. Alnemri, E.S., et al., Human ICE/CED-3 protease nomenclature. Cell, 1996. 87(2): p. 171. Cohen, G.M., Caspases: the executioners of apoptosis. Biochem J, 1997. 326 ( Pt 1): p. 1-16. Hengartner, M.O., The biochemistry of apoptosis. Nature, 2000. 407(6805): p. 770-6. Crawford, E.D. and J.A. Wells, Caspase substrates and cellular remodeling. Annu Rev Biochem, 2011. 80: p. 1055-87. Buendia, B., A. Santa-Maria, and J.C. Courvalin, Caspase-dependent proteolysis of integral and peripheral proteins of nuclear membranes and nuclear pore complex proteins during apoptosis. J Cell Sci, 1999. 112 ( Pt 11): p. 1743-53. Rao, L., D. Perez, and E. White, Lamin proteolysis facilitates nuclear events during apoptosis. J Cell Biol, 1996. 135(6 Pt 1): p. 1441-55. Nagata, S., Apoptotic DNA fragmentation. Exp Cell Res, 2000. 256(1): p. 128. 194 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. Elmore, S., Apoptosis: a review of programmed cell death. Toxicol Pathol, 2007. 35(4): p. 495-516. Hsu, H., J. Xiong, and D.V. Goeddel, The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell, 1995. 81(4): p. 495-504. Wajant, H., The Fas signaling pathway: more than a paradigm. Science, 2002. 296(5573): p. 1635-6. Kischkel, F.C., et al., Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J, 1995. 14(22): p. 5579-88. Ozoren, N. and W.S. El-Deiry, Defining characteristics of Types I and II apoptotic cells in response to TRAIL. Neoplasia, 2002. 4(6): p. 551-7. Scaffidi, C., et al., Two CD95 (APO-1/Fas) signaling pathways. EMBO J, 1998. 17(6): p. 1675-87. Kroemer, G., L. Galluzzi, and C. Brenner, Mitochondrial membrane permeabilization in cell death. Physiol Rev, 2007. 87(1): p. 99-163. Chinnaiyan, A.M., The apoptosome: heart and soul of the cell death machine. Neoplasia, 1999. 1(1): p. 5-15. Li, P., et al., Cytochrome c and dATP-dependent formation of Apaf-1/caspase9 complex initiates an apoptotic protease cascade. Cell, 1997. 91(4): p. 47989. Cain, K., et al., Caspase activation involves the formation of the aposome, a large (approximately 700 kDa) caspase-activating complex. J Biol Chem, 1999. 274(32): p. 22686-92. Stennicke, H.R., et al., Caspase-9 can be activated without proteolytic processing. J Biol Chem, 1999. 274(13): p. 8359-62. Schimmer, A.D., Inhibitor of apoptosis proteins: translating basic knowledge into clinical practice. Cancer Res, 2004. 64(20): p. 7183-90. van Loo, G., et al., The serine protease Omi/HtrA2 is released from mitochondria during apoptosis. Omi interacts with caspase-inhibitor XIAP and induces enhanced caspase activity. Cell Death Differ, 2002. 9(1): p. 20-6. Adams, J.M. and S. Cory, The Bcl-2 protein family: arbiters of cell survival. Science, 1998. 281(5381): p. 1322-6. Gross, A., J.M. McDonnell, and S.J. Korsmeyer, BCL-2 family members and the mitochondria in apoptosis. Genes Dev, 1999. 13(15): p. 1899-911. Reed, J.C., Double identity for proteins of the Bcl-2 family. Nature, 1997. 387(6635): p. 773-6. Cleary, M.L., S.D. Smith, and J. Sklar, Cloning and structural analysis of cDNAs for bcl-2 and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation. Cell, 1986. 47(1): p. 19-28. Nguyen, M., et al., Targeting of Bcl-2 to the mitochondrial outer membrane by a COOH-terminal signal anchor sequence. J Biol Chem, 1993. 268(34): p. 25265-8. Lithgow, T., et al., The protein product of the oncogene bcl-2 is a component of the nuclear envelope, the endoplasmic reticulum, and the outer mitochondrial membrane. Cell Growth Differ, 1994. 5(4): p. 411-7. Petros, A.M., E.T. Olejniczak, and S.W. Fesik, Structural biology of the Bcl-2 family of proteins. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 2004. 1644(2–3): p. 83-94. 195 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. Petros, A.M., et al., Solution structure of the antiapoptotic protein bcl-2. Proceedings of the National Academy of Sciences, 2001. 98(6): p. 3012-3017. Basu, A., S.A. You, and S. Haldar, Regulation of Bcl2 phosphorylation by stress response kinase pathway. Int J Oncol, 2000. 16(3): p. 497-500. Bassik, M.C., et al., Phosphorylation of BCL-2 regulates ER Ca2+ homeostasis and apoptosis. EMBO J, 2004. 23(5): p. 1207-16. Deng, X., et al., Mono- and multisite phosphorylation enhances Bcl2's antiapoptotic function and inhibition of cell cycle entry functions. Proc Natl Acad Sci U S A, 2004. 101(1): p. 153-8. Ito, T., et al., Bcl-2 phosphorylation required for anti-apoptosis function. J Biol Chem, 1997. 272(18): p. 11671-3. Vaux, D.L., S. Cory, and J.M. Adams, Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature, 1988. 335(6189): p. 440-2. Strasser, A., et al., Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature, 1990. 348(6299): p. 3313. Low, I.C., J. Kang, and S. Pervaiz, Bcl-2: a prime regulator of mitochondrial redox metabolism in cancer cells. Antioxid Redox Signal, 2011. 15(12): p. 2975-87. Chipuk, J.E., et al., The BCL-2 family reunion. Mol Cell, 2010. 37(3): p. 299310. Mikhailov, V., et al., Association of Bax and Bak homo-oligomers in mitochondria. Bax requirement for Bak reorganization and cytochrome c release. J Biol Chem, 2003. 278(7): p. 5367-76. Kuwana, T., et al., Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane. Cell, 2002. 111(3): p. 331342. Wei, M.C., et al., tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c. Genes and Development, 2000. 14(16): p. 20602071. Kuwana, T., et al., BH3 Domains of BH3-Only Proteins Differentially Regulate Bax-Mediated Mitochondrial Membrane Permeabilization Both Directly and Indirectly. Mol Cell, 2005. 17(4): p. 525-535. Letai, A., et al., Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell, 2002. 2(3): p. 183-192. Uren, R.T., et al., Mitochondrial permeabilization relies on BH3 ligands engaging multiple prosurvival Bcl-2 relatives, not Bak. Journal of Cell Biology, 2007. 177(2): p. 277-287. Willis, S.N., et al., Apoptosis initiated when BH3 ligands engage multiple Bcl2 homologs, not Bax or Bak. Science, 2007. 315(5813): p. 856-859. Wei, M.C., et al., Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science, 2001. 292(5517): p. 727-30. Ojala, P.M., et al., The apoptotic v-cyclin-CDK6 complex phosphorylates and inactivates Bcl-2. Nat Cell Biol, 2000. 2(11): p. 819-25. Yamamoto, K., H. Ichijo, and S.J. Korsmeyer, BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G(2)/M. Mol Cell Biol, 1999. 19(12): p. 8469-78. 196 152. 153. 154. 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. Nencioni, L., et al., Bcl-2 expression and p38MAPK activity in cells infected with influenza A virus: impact on virally induced apoptosis and viral replication. J Biol Chem, 2009. 284(23): p. 16004-15. Thomas, A., T. Giesler, and E. White, p53 mediates bcl-2 phosphorylation and apoptosis via activation of the Cdc42/JNK1 pathway. Oncogene, 2000. 19(46): p. 5259-69. De Chiara, G., et al., Bcl-2 Phosphorylation by p38 MAPK: identification of target sites and biologic consequences. J Biol Chem, 2006. 281(30): p. 2135361. Kang, Y.H. and S.J. Lee, The role of p38 MAPK and JNK in Arsenic trioxideinduced mitochondrial cell death in human cervical cancer cells. J Cell Physiol, 2008. 217(1): p. 23-33. Wei, Y., et al., JNK1-mediated phosphorylation of Bcl-2 regulates starvationinduced autophagy. Mol Cell, 2008. 30(6): p. 678-88. Liu, Y., et al., Activation of ERK-p53 and ERK-Mediated Phosphorylation of Bcl-2 Are Involved in Autophagic Cell Death Induced by the c-Met Inhibitor SU11274 in Human Lung Cancer A549 Cells. J Pharmacol Sci, 2012. 118(4): p. 423-32. Deng, X., et al., Survival function of ERK1/2 as IL-3-activated, staurosporineresistant Bcl2 kinases. Proc Natl Acad Sci U S A, 2000. 97(4): p. 1578-83. May, W.S., et al., Interleukin-3 and bryostatin-1 mediate hyperphosphorylation of BCL2 alpha in association with suppression of apoptosis. J Biol Chem, 1994. 269(43): p. 26865-70. Asnaghi, L., et al., Bcl-2 phosphorylation and apoptosis activated by damaged microtubules require mTOR and are regulated by Akt. Oncogene, 2004. 23(34): p. 5781-91. Pathan, N., et al., Microtubule-targeting drugs induce bcl-2 phosphorylation and association with Pin1. Neoplasia, 2001. 3(6): p. 550-9. Boudny, V. and S. Nakano, Src tyrosine kinase augments taxotere-induced apoptosis through enhanced expression and phosphorylation of Bcl-2. Br J Cancer, 2002. 86(3): p. 463-9. Lin, S.S., et al., PP2A regulates BCL-2 phosphorylation and proteasomemediated degradation at the endoplasmic reticulum. J Biol Chem, 2006. 281(32): p. 23003-12. Ruvolo, P.P., et al., Ceramide induces Bcl2 dephosphorylation via a mechanism involving mitochondrial PP2A. J Biol Chem, 1999. 274(29): p. 20296-300. Brichese, L. and A. Valette, PP1 phosphatase is involved in Bcl-2 dephosphorylation after prolonged mitotic arrest induced by paclitaxel. Biochemical and Biophysical Research Communications, 2002. 294(2): p. 504-508. Srivastava, R.K., et al., Deletion of the loop region of Bcl-2 completely blocks paclitaxel-induced apoptosis. Proc Natl Acad Sci U S A, 1999. 96(7): p. 377580. Haldar, S., J. Chintapalli, and C.M. Croce, Taxol induces bcl-2 phosphorylation and death of prostate cancer cells. Cancer Res, 1996. 56(6): p. 1253-5. Konopleva, M., et al., Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell, 2006. 10(5): p. 375-88. 197 169. 170. 171. 172. 173. 174. 175. 176. 177. 178. 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. Shitashige, M., et al., Dissociation of Bax from a Bcl-2/Bax heterodimer triggered by phosphorylation of serine 70 of Bcl-2. J Biochem, 2001. 130(6): p. 741-8. Mai, H., et al., A functional role for nicotine in Bcl2 phosphorylation and suppression of apoptosis. J Biol Chem, 2003. 278(3): p. 1886-91. Ray, R.M., S. Bhattacharya, and L.R. Johnson, Protein phosphatase 2A regulates apoptosis in intestinal epithelial cells. J Biol Chem, 2005. 280(35): p. 31091-100. Ruvolo, P.P., X. Deng, and W.S. May, Phosphorylation of Bcl2 and regulation of apoptosis. Leukemia, 2001. 15(4): p. 515-22. Srivastava, R.K., et al., Involvement of microtubules in the regulation of Bcl2 phosphorylation and apoptosis through cyclic AMP-dependent protein kinase. Mol Cell Biol, 1998. 18(6): p. 3509-17. Fridlyand, L.E. and L.H. Philipson, Reactive species and early manifestation of insulin resistance in type diabetes. Diabetes Obes Metab, 2006. 8(2): p. 136-45. Hool, L.C., Reactive oxygen species in cardiac signalling: from mitochondria to plasma membrane ion channels. Clin Exp Pharmacol Physiol, 2006. 33(12): p. 146-51. Larbi, A., J. Kempf, and G. Pawelec, Oxidative stress modulation and T cell activation. Exp Gerontol, 2007. 42(9): p. 852-8. Piccoli, C., et al., Role of reactive oxygen species as signal molecules in the pre-commitment phase of adult stem cells. Ital J Biochem, 2007. 56(4): p. 295301. Touyz, R.M., F. Tabet, and E.L. Schiffrin, Redox-dependent signalling by angiotensin II and vascular remodelling in hypertension. Clin Exp Pharmacol Physiol, 2003. 30(11): p. 860-6. Pervaiz, S. and M.V. Clement, Superoxide anion: oncogenic reactive oxygen species? Int J Biochem Cell Biol, 2007. 39(7-8): p. 1297-304. Armstrong, A.W., et al., Angiogenesis and oxidative stress: common mechanisms linking psoriasis with atherosclerosis. J Dermatol Sci, 2011. 63(1): p. 1-9. Jomova, K., et al., Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem, 2010. 345(1-2): p. 91-104. Park, J.G. and G.T. Oh, The role of peroxidases in the pathogenesis of atherosclerosis. BMB Rep, 2011. 44(8): p. 497-505. Taupin, P., A dual activity of ROS and oxidative stress on adult neurogenesis and Alzheimer's disease. Cent Nerv Syst Agents Med Chem, 2010. 10(1): p. 16-21. Victor, V.M., et al., Oxidative stress and mitochondrial dysfunction in type diabetes. Curr Pharm Des, 2011. 17(36): p. 3947-58. Ushio-Fukai, M. and Y. Nakamura, Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett, 2008. 266(1): p. 37-52. Pani, G., T. Galeotti, and P. Chiarugi, Metastasis: cancer cell's escape from oxidative stress. Cancer Metastasis Rev, 2010. 29(2): p. 351-78. Li, Z.Y., et al., Mitochondrial ROS generation for regulation of autophagic pathways in cancer. Biochem Biophys Res Commun, 2011. 414(1): p. 5-8. Saito, Y., et al., Turning point in apoptosis/necrosis induced by hydrogen peroxide. Free Radic Res, 2006. 40(6): p. 619-30. 198 189. 190. 191. 192. 193. 194. 195. 196. 197. 198. 199. 200. 201. 202. 203. 204. 205. Takeda, M., et al., Hydrogen peroxide induces necrosis, apoptosis, oncosis and apoptotic oncosis of mouse terminal proximal straight tubule cells. Nephron, 1999. 81(2): p. 234-8. Teramoto, S., et al., Hydrogen peroxide-induced apoptosis and necrosis in human lung fibroblasts: protective roles of glutathione. Jpn J Pharmacol, 1999. 79(1): p. 33-40. Ahmad, K.A., et al., Hydrogen peroxide-mediated cytosolic acidification is a signal for mitochondrial translocation of Bax during drug-induced apoptosis of tumor cells. Cancer Res, 2004. 64(21): p. 7867-78. Inoshita, S., et al., Phosphorylation and inactivation of myeloid cell leukemia by JNK in response to oxidative stress. J Biol Chem, 2002. 277(46): p. 43730-4. Schroeter, H., et al., c-Jun N-terminal kinase (JNK)-mediated modulation of brain mitochondria function: New target proteins for JNK signalling in mitochondrion-dependent apoptosis. Biochemical Journal, 2003. 372(2): p. 359-369. Naughton, R., et al., Bcr-Abl-mediated redox regulation of the PI3K/AKT pathway. Leukemia, 2009. 23(8): p. 1432-1440. Leslie, N.R., et al., Redox regulation of PI 3-kinase signalling via inactivation of PTEN. EMBO Journal, 2003. 22(20): p. 5501-5510. Shibanuma, M., T. Kuroki, and K. Nose, Induction of DNA replication and expression of proto-oncogene c-myc and c-fos in quiescent Balb/3T3 cells by xanthine/xanthine oxidase. Oncogene, 1988. 3(1): p. 17-21. Burdon, R.H., Superoxide and hydrogen peroxide in relation to mammalian cell proliferation. Free Radical Biology and Medicine, 1995. 18(4): p. 775794. Pervaiz, S., et al., Superoxide anion inhibits drug-induced tumor cell death. FEBS Lett, 1999. 459(3): p. 343-8. Clement, M.V. and I. Stamenkovic, Superoxide anion is a natural inhibitor of FAS-mediated cell death. EMBO J, 1996. 15(2): p. 216-25. Clement, M.V., J.L. Hirpara, and S. Pervaiz, Decrease in intracellular superoxide sensitizes Bcl-2-overexpressing tumor cells to receptor and druginduced apoptosis independent of the mitochondria. Cell Death Differ, 2003. 10(11): p. 1273-85. Clément, M.V. and S. Pervaiz, Intracellular superoxide and hydrogen peroxide concentrations: A critical balance that determines survival or death. Redox Report, 2001. 6(4): p. 211-214. Zhu, P., et al., Angiopoietin-like Protein Elevates the Prosurvival Intracellular O2−:H2O2 Ratio and Confers Anoikis Resistance to Tumors. Cancer Cell, 2011. 19(3): p. 401-415. Clement, M.V., A. Ponton, and S. Pervaiz, Apoptosis induced by hydrogen peroxide is mediated by decreased superoxide anion concentration and reduction of intracellular milieu. FEBS Lett, 1998. 440(1-2): p. 13-8. Hirpara, J.L., M.V. Clement, and S. Pervaiz, Intracellular acidification triggered by mitochondrial-derived hydrogen peroxide is an effector mechanism for drug-induced apoptosis in tumor cells. J Biol Chem, 2001. 276(1): p. 514-21. Barbosa, L.F., et al., Increased SOD1 association with chromatin, DNA damage, p53 activation, and apoptosis in a cellular model of SOD1-linked 199 206. 207. 208. 209. 210. 211. 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. ALS. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 2010. 1802(5): p. 462-471. Zhang, Y., et al., Overexpression of copper zinc superoxide dismutase suppresses human glioma cell growth. Cancer Res, 2002. 62(4): p. 1205-1212. Lin, K.I., et al., Decreased intracellular superoxide levels activate Sindbis virus-induced apoptosis. J Biol Chem, 1999. 274(19): p. 13650-5. Chen, Z.X. and S. Pervaiz, Bcl-2 induces pro-oxidant state by engaging mitochondrial respiration in tumor cells. Cell Death Differ, 2007. 14(9): p. 1617-27. Chen, Z.X. and S. Pervaiz, Involvement of cytochrome c oxidase subunits Va and Vb in the regulation of cancer cell metabolism by Bcl-2. Cell Death Differ, 2010. 17(3): p. 408-20. Virshup, D.M. and S. Shenolikar, From promiscuity to precision: protein phosphatases get a makeover. Mol Cell, 2009. 33(5): p. 537-45. Xu, Y., et al., Structure of the protein phosphatase 2A holoenzyme. Cell, 2006. 127(6): p. 1239-51. Eichhorn, P.J., M.P. Creyghton, and R. Bernards, Protein phosphatase 2A regulatory subunits and cancer. Biochim Biophys Acta, 2009. 1795(1): p. 115. Stone, S.R., J. Hofsteenge, and B.A. Hemmings, Molecular cloning of cDNAs encoding two isoforms of the catalytic subunit of protein phosphatase 2A. Biochemistry, 1987. 26(23): p. 7215-20. Hemmings, B.A., et al., alpha- and beta-forms of the 65-kDa subunit of protein phosphatase 2A have a similar 39 amino acid repeating structure. Biochemistry, 1990. 29(13): p. 3166-73. McCright, B., et al., The B56 family of protein phosphatase 2A (PP2A) regulatory subunits encodes differentiation-induced phosphoproteins that target PP2A to both nucleus and cytoplasm. J Biol Chem, 1996. 271(36): p. 22081-9. Yang, J. and C. Phiel, Functions of B56-containing PP2As in major developmental and cancer signaling pathways. Life Sci, 2011. 87(23-26): p. 659-66. Moreno, C.S., et al., WD40 repeat proteins striatin and S/G(2) nuclear autoantigen are members of a novel family of calmodulin-binding proteins that associate with protein phosphatase 2A. J Biol Chem, 2000. 275(8): p. 5257-63. Stark, M.J., Yeast protein serine/threonine phosphatases: multiple roles and diverse regulation. Yeast, 1996. 12(16): p. 1647-75. Götz, J., et al., Delayed embryonic lethality in mice lacking protein phosphatase 2A catalytic subunit Cα. Proc Natl Acad Sci U S A, 1998. 95(21): p. 12370-12375. Strack, S., J.T. Cribbs, and L. Gomez, Critical role for protein phosphatase 2A heterotrimers in mammalian cell survival. J Biol Chem, 2004. 279(46): p. 47732-9. Janssens, V. and J. Goris, Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem J, 2001. 353(Pt 3): p. 417-39. Wurzenberger, C. and D.W. Gerlich, Phosphatases: providing safe passage through mitotic exit. Nat Rev Mol Cell Biol, 2011. 12(8): p. 469-82. 200 223. 224. 225. 226. 227. 228. 229. 230. 231. 232. 233. 234. 235. 236. 237. 238. 239. 240. 241. 242. Collins, E. and A.T. Sim, Regulation of neuronal PP1 and PP2A during development. Methods Mol Biol, 1998. 93: p. 79-102. Basu, S., PP2A in the regulation of cell motility and invasion. Curr Protein Pept Sci, 2011. 12(1): p. 3-11. Suganuma, M., et al., Okadaic acid: an additional non-phorbol-12tetradecanoate-13-acetate-type tumor promoter. Proc Natl Acad Sci U S A, 1988. 85(6): p. 1768-71. Pallas, D.C., et al., Polyoma small and middle T antigens and SV40 small t antigen form stable complexes with protein phosphatase 2A. Cell, 1990. 60(1): p. 167-76. Campbell, K.S., et al., Identification of regions in polyomavirus middle T and small t antigens important for association with protein phosphatase 2A. J Virol, 1995. 69(6): p. 3721-8. Wang, S.S., et al., Alterations of the PPP2R1B gene in human lung and colon cancer. Science, 1998. 282(5387): p. 284-7. Campbell, I.G. and T. Manolitsas, Absence of PPP2R1B gene alterations in primary ovarian cancers. Oncogene, 1999. 18(46): p. 6367-9. Takagi, Y., et al., Alterations of the PPP2R1B gene located at 11q23 in human colorectal cancers. Gut, 2000. 47(2): p. 268-71. Calin, G.A., et al., Low frequency of alterations of the alpha (PPP2R1A) and beta (PPP2R1B) isoforms of the subunit A of the serine-threonine phosphatase 2A in human neoplasms. Oncogene, 2000. 19(9): p. 1191-5. Arnold, H.K. and R.C. Sears, Protein phosphatase 2A regulatory subunit B56alpha associates with c-myc and negatively regulates c-myc accumulation. Mol Cell Biol, 2006. 26(7): p. 2832-44. Felsher, D.W. and J.M. Bishop, Reversible Tumorigenesis by MYC in Hematopoietic Lineages. Mol Cell, 1999. 4(2): p. 199-207. Pelengaris, S., et al., Reversible Activation of c-Myc in Skin: Induction of a Complex Neoplastic Phenotype by a Single Oncogenic Lesion. Mol Cell, 1999. 3(5): p. 565-577. Magenta, A., et al., Protein phosphatase 2A subunit PR70 interacts with pRb and mediates its dephosphorylation. Mol Cell Biol, 2008. 28(2): p. 873-82. Cobrinik, D., Pocket proteins and cell cycle control. 2005. 24(17): p. 27962809. Mittnacht, S., The retinoblastoma protein—from bench to bedside. European Journal of Cell Biology, 2005. 84(2–3): p. 97-107. Li, H.H., et al., A specific PP2A regulatory subunit, B56gamma, mediates DNA damage-induced dephosphorylation of p53 at Thr55. EMBO J, 2007. 26(2): p. 402-11. Letourneux, C., G. Rocher, and F. Porteu, B56-containing PP2A dephosphorylate ERK and their activity is controlled by the early gene IEX-1 and ERK. EMBO J, 2006. 25(4): p. 727-38. Pearson, G., et al., Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev, 2001. 22(2): p. 153-83. Foley, T.D., J.J. Armstrong, and B.R. Kupchak, Identification and H2O2 sensitivity of the major constitutive MAPK phosphatase from rat brain. Biochem Biophys Res Commun, 2004. 315(3): p. 568-74. Chen, L., L. Liu, and S. Huang, Cadmium activates the mitogen-activated protein kinase (MAPK) pathway via induction of reactive oxygen species and 201 243. 244. 245. 246. 247. 248. 249. 250. 251. 252. 253. 254. 255. 256. 257. 258. inhibition of protein phosphatases 2A and 5. Free Radic Biol Med, 2008. 45(7): p. 1035-44. Ruvolo, P.P., et al., A functional role for the B56 alpha-subunit of protein phosphatase 2A in ceramide-mediated regulation of Bcl2 phosphorylation status and function. J Biol Chem, 2002. 277(25): p. 22847-52. Acharya, A., et al., Redox regulation in cancer: a double-edged sword with therapeutic potential. Oxid Med Cell Longev, 2010. 3(1): p. 23-34. Hsu, W.H., et al., Berberine induces apoptosis in SW620 human colonic carcinoma cells through generation of reactive oxygen species and activation of JNK/p38 MAPK and FasL. Arch Toxicol, 2007. 81(10): p. 719-28. Kim, M.H., et al., Acetylcholine inhibits long-term hypoxia-induced apoptosis by suppressing the oxidative stress-mediated MAPKs activation as well as regulation of Bcl-2, c-IAPs, and caspase-3 in mouse embryonic stem cells. Apoptosis, 2008. 13(2): p. 295-304. Kuo, P.L., et al., Involvement of reactive oxygen species/c-Jun NH(2)-terminal kinase pathway in kotomolide A induces apoptosis in human breast cancer cells. Toxicol Appl Pharmacol, 2008. 229(2): p. 215-26. Shim, H.Y., et al., Acacetin-induced apoptosis of human breast cancer MCF-7 cells involves caspase cascade, mitochondria-mediated death signaling and SAPK/JNK1/2-c-Jun activation. Mol Cells, 2007. 24(1): p. 95-104. Kim, H.S., et al., Constitutive induction of p-Erk1/2 accompanied by reduced activities of protein phosphatases and 2A and MKP3 due to reactive oxygen species during cellular senescence. J Biol Chem, 2003. 278(39): p. 37497510. Banker, D.E., et al., Measurement of spontaneous and therapeutic agentinduced apoptosis with BCL-2 protein expression in acute myeloid leukemia. Blood, 1997. 89(1): p. 243-55. Campos, L., et al., Expression of apoptosis-controlling proteins in acute leukemia cells. Leuk Lymphoma, 1999. 33(5-6): p. 499-509. Lickliter, J.D., et al., HA14-1 selectively induces apoptosis in Bcl-2overexpressing leukemia/lymphoma cells, and enhances cytarabine-induced cell death. Leukemia, 2003. 17(11): p. 2074-80. Skinnider, B.F., et al., Bcl-6 and Bcl-2 protein expression in diffuse large Bcell lymphoma and follicular lymphoma: correlation with 3q27 and 18q21 chromosomal abnormalities. Hum Pathol, 1999. 30(7): p. 803-8. Huang, S.T. and J.A. Cidlowski, Phosphorylation status modulates Bcl-2 function during glucocorticoid-induced apoptosis in T lymphocytes. FASEB J, 2002. 16(8): p. 825-32. Wright, V.P., et al., Superoxide scavengers augment contractile but not energetic responses to hypoxia in rat diaphragm. Journal of Applied Physiology, 2005. 98(5): p. 1753-1760. Tilly, J.L. and K.I. Tilly, Inhibitors of oxidative stress mimic the ability of follicle-stimulating hormone to suppress apoptosis in cultured rat ovarian follicles. Endocrinology, 1995. 136(1): p. 242-52. Davies, C.M., et al., Reactive nitrogen and oxygen species in interleukin-1mediated DNA damage associated with osteoarthritis. Osteoarthritis Cartilage, 2008. 16(5): p. 624-30. Low, I.C., Z.X. Chen, and S. Pervaiz, Bcl-2 modulates resveratrol-induced ROS production by regulating mitochondrial respiration in tumor cells. Antioxid Redox Signal, 2010. 13(6): p. 807-19. 202 259. 260. 261. 262. 263. 264. 265. 266. 267. 268. 269. 270. 271. 272. 273. 274. 275. 276. 277. Karpinich, N.O., et al., The course of etoposide-induced apoptosis in Jurkat cells lacking p53 and Bax. J Cell Physiol, 2006. 208(1): p. 55-63. Li, X., et al., B56-associated protein phosphatase 2A is required for survival and protects from apoptosis in Drosophila melanogaster. Mol Cell Biol, 2002. 22(11): p. 3674-84. Chen, F., et al., Multiple protein phosphatases are required for mitosis in Drosophila. Curr Biol, 2007. 17(4): p. 293-303. Csortos, C., et al., High complexity in the expression of the B' subunit of protein phosphatase 2A0. Evidence for the existence of at least seven novel isoforms. J Biol Chem, 1996. 271(5): p. 2578-88. McCright, B. and D.M. Virshup, Identification of a new family of protein phosphatase 2A regulatory subunits. J Biol Chem, 1995. 270(44): p. 26123-8. Lin, K.T., et al., Peroxynitrite induces apoptosis of HL-60 cells by activation of a caspase-3 family protease. Am J Physiol, 1998. 274(4 Pt 1): p. C855-60. Sandoval, M., et al., Peroxynitrite-induced apoptosis in T84 and RAW 264.7 cells: attenuation by L-ascorbic acid. Free Radic Biol Med, 1997. 22(3): p. 489-95. Wu, F. and J.X. Wilson, Peroxynitrite-dependent activation of protein phosphatase type 2A mediates microvascular endothelial barrier dysfunction. Cardiovasc Res, 2009. 81(1): p. 38-45. Li, X. and D.M. Virshup, Two conserved domains in regulatory B subunits mediate binding to the A subunit of protein phosphatase 2A. Eur J Biochem, 2002. 269(2): p. 546-52. Souza, J.M., et al., Factors determining the selectivity of protein tyrosine nitration. Arch Biochem Biophys, 1999. 371(2): p. 169-78. Sacksteder, C.A., et al., Endogenously nitrated proteins in mouse brain: links to neurodegenerative disease. Biochemistry, 2006. 45(26): p. 8009-22. Forester, C.M., et al., Control of mitotic exit by PP2A regulation of Cdc25C and Cdk1. Proc Natl Acad Sci U S A, 2007. 104(50): p. 19867-72. Louis, J.V., et al., Mice lacking phosphatase PP2A subunit PR61/B'delta (Ppp2r5d) develop spatially restricted tauopathy by deregulation of CDK5 and GSK3beta. Proc Natl Acad Sci U S A, 2011. 108(17): p. 6957-62. Adams, D.G., et al., Positive regulation of Raf1-MEK1/2-ERK1/2 signaling by protein serine/threonine phosphatase 2A holoenzymes. J Biol Chem, 2005. 280(52): p. 42644-54. Giannoni, E., et al., Redox regulation of anoikis resistance of metastatic prostate cancer cells: key role for Src and EGFR-mediated pro-survival signals. Oncogene, 2009. 28(20): p. 2074-86. Chiarugi, P. and E. Giannoni, Anchorage-dependent cell growth: tyrosine kinases and phosphatases meet redox regulation. Antioxid Redox Signal, 2005. 7(5-6): p. 578-92. Irani, K. and P.J. Goldschmidt-Clermont, Ras, superoxide and signal transduction. Biochem Pharmacol, 1998. 55(9): p. 1339-46. Narayanan, A., et al., Alteration in superoxide dismutase causes oxidative stress and p38 MAPK activation following RVFV infection. PLoS One, 2011. 6(5): p. e20354. Somwar, R., et al., Superoxide dismutase (SOD1) is a target for a small molecule identified in a screen for inhibitors of the growth of lung adenocarcinoma cell lines. Proceedings of the National Academy of Sciences, 2011. 108(39): p. 16375-16380. 203 278. 279. 280. 281. 282. 283. 284. 285. 286. 287. 288. 289. 290. 291. 292. 293. 294. 295. Brown, D.P., et al., Targeting superoxide dismutase to overcome cisplatin resistance in human ovarian cancer. Cancer Chemother Pharmacol, 2009. 63(4): p. 723-30. Huang, P., et al., Superoxide dismutase as a target for the selective killing of cancer cells. Nature, 2000. 407(6802): p. 390-395. Yoshizaki, N., et al., Suppressive effect of recombinant human Cu, Znsuperoxide dismutase on lung metastasis of murine tumor cells. International Journal of Cancer, 1994. 57(2): p. 287-292. Kogawa, K., et al., Enhanced inhibition of experimental metastasis by the combination chemotherapy of Cu-Zn SOD and adriamycin. Clinical and Experimental Metastasis, 1999. 17(3): p. 239-244. Nonaka, Y., et al., Effect of reactive oxygen intermediates on the in vitro invasive capacity of tumor cells and liver metastasis in mice. International Journal of Cancer, 1993. 54(6): p. 983-986. Van Driel, B.E.M., et al., Expression of CuZn- and Mn-Superoxide Dismutase in Human Colorectal Neoplasms. Free Radical Biology and Medicine, 1997. 23(3): p. 435-444. Oberley, T.D., et al., Immunogold analysis of antioxidant enzymes in human renal cell carcinoma. Virchows Archiv, 1994. 424(2): p. 155-164. Oberley, T.D., J.M. Sempf, and L.W. Oberley, Immunogold analysis of antioxidant enzymes in common renal cancers. Histology and Histopathology, 1996. 11(1): p. 153-160. Baker, A.M., L.W. Oberley, and M.B. Cohen, Expression of antioxidant enzymes in human prostatic adenocarcinoma. Prostate, 1997. 32(4): p. 229233. Bostwick, D.G., et al., Antioxidant enzyme expression and reactive oxygen species damage in prostatic intraepithelial neoplasia and cancer. Cancer, 2000. 89(1): p. 123-134. Cullen, J.J., F.A. Mitros, and L.W. Oberley, Expression of antioxidant enzymes in diseases of the human pancreas: Another link between chronic pancreatitis and pancreatic cancer. Pancreas, 2003. 26(1): p. 23-27. Coursin, D.B., et al., An immunohistochemical analysis of antioxidant and glutathione S-transferase enzyme levels in normal and neoplastic human lung. Histology and Histopathology, 1996. 11(4): p. 851-860. Janssen, A.M.L., et al., Superoxide dismutases in relation to the overall survival of colorectal cancer patients. Br J Cancer, 1998. 78(8): p. 1051-1057. Bianchi, M.S., N.O. Bianchi, and A.D. Bolzán, Superoxide dismutase activity and superoxide dismutase-1 gene methylation in normal and tumoral human breast tissues. Cancer Genetics and Cytogenetics, 1992. 59(1): p. 26-29. Gorg, B., et al., Reversible inhibition of mammalian glutamine synthetase by tyrosine nitration. FEBS Lett, 2007. 581(1): p. 84-90. Kamisaki, Y., et al., An activity in rat tissues that modifies nitrotyrosinecontaining proteins. Proc Natl Acad Sci U S A, 1998. 95(20): p. 11584-9. Smallwood, H.S., et al., Identification of a denitrase activity against calmodulin in activated macrophages using high-field liquid chromatography-FTICR mass spectrometry. Biochemistry, 2007. 46(37): p. 10498-505. Irie, Y., et al., Histone H1.2 is a substrate for denitrase, an activity that reduces nitrotyrosine immunoreactivity in proteins. Proc Natl Acad Sci U S A, 2003. 100(10): p. 5634-9. 204 296. 297. 298. 299. 300. 301. 302. 303. 304. 305. 306. 307. 308. 309. 310. 311. 312. 313. 314. Kang, M. and H.I. Akbarali, Denitration of L-type calcium channel. FEBS Lett, 2008. 582(20): p. 3033-6. Lambeth, J.D., NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol, 2004. 4(3): p. 181-9. Bae, Y.S., et al., Regulation of reactive oxygen species generation in cell signaling. Mol Cells, 2011. 32(6): p. 491-509. Perner, A., et al., Superoxide production and expression of NAD(P)H oxidases by transformed and primary human colonic epithelial cells. Gut, 2003. 52(2): p. 231-6. Brar, S.S., et al., NOX5 NAD(P)H oxidase regulates growth and apoptosis in DU 145 prostate cancer cells. Am J Physiol Cell Physiol, 2003. 285(2): p. C353-69. Brar, S.S., et al., An NAD(P)H oxidase regulates growth and transcription in melanoma cells. Am J Physiol Cell Physiol, 2002. 282(6): p. C1212-24. Geiszt, M., et al., NAD(P)H oxidase 1, a product of differentiated colon epithelial cells, can partially replace glycoprotein 91phox in the regulated production of superoxide by phagocytes. J Immunol, 2003. 171(1): p. 299306. Hilenski, L.L., et al., Distinct subcellular localizations of Nox1 and Nox4 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol, 2004. 24(4): p. 677-83. Brand, M.D., The sites and topology of mitochondrial superoxide production. Exp Gerontol, 2010. 45(7-8): p. 466-72. Madesh, M. and G. Hajnóczky, VDAC-dependent permeabilization of the outer mitochondrial membrane by superoxide induces rapid and massive cytochrome c release. J Cell Biol, 2001. 155(6): p. 1003-1016. Han, D., et al., Voltage-dependent anion channels control the release of the superoxide anion from mitochondria to cytosol. J Biol Chem, 2003. 278(8): p. 5557-63. Vakkala, M., et al., Inducible nitric oxide synthase expression, apoptosis, and angiogenesis in in situ and invasive breast carcinomas. Clin Cancer Res, 2000. 6(6): p. 2408-16. Ambs, S., et al., Relationship between p53 mutations and inducible nitric oxide synthase expression in human colorectal cancer. J Natl Cancer Inst, 1999. 91(1): p. 86-8. Tanaka, H., et al., Frequent expression of inducible nitric oxide synthase in esophageal squamous cell carcinomas. Int J Oncol, 1999. 14(6): p. 1069-73. Yamaguchi, S., et al., Glioma tumourgenicity is decreased by iNOS knockout: experimental studies using the C6 striatal implantation glioma model. Br J Neurosurg, 2002. 16(6): p. 567-72. Jenkins, D.C., et al., Roles of nitric oxide in tumor growth. Proc Natl Acad Sci U S A, 1995. 92(10): p. 4392-6. Barreiro Arcos, M.L., et al., Inducible nitric oxide synthase-mediated proliferation of a T lymphoma cell line. Nitric Oxide, 2003. 8(2): p. 111-8. Shi, Q., et al., Direct correlation between nitric oxide synthase II inducibility and metastatic ability of UV-2237 murine fibrosarcoma cells carrying mutant p53. Cancer Res, 1999. 59(9): p. 2072-5. Le, X., et al., Nitric oxide synthase II suppresses the growth and metastasis of human cancer regardless of its up-regulation of protumor factors. Proc Natl Acad Sci U S A, 2005. 102(24): p. 8758-63. 205 315. 316. 317. 318. 319. 320. 321. 322. 323. 324. 325. 326. 327. 328. 329. 330. 331. 332. Wang, B., et al., A novel model system for studying the double-edged roles of nitric oxide production in pancreatic cancer growth and metastasis. Oncogene, 2003. 22(12): p. 1771-82. Marchenko, N.D., A. Zaika, and U.M. Moll, Death signal-induced localization of p53 protein to mitochondria. A potential role in apoptotic signaling. J Biol Chem, 2000. 275(21): p. 16202-12. Mihara, M., et al., p53 has a direct apoptogenic role at the mitochondria. Mol Cell, 2003. 11(3): p. 577-90. Deng, X., F. Gao, and W.S. May, Protein phosphatase 2A inactivates Bcl2's antiapoptotic function by dephosphorylation and up-regulation of Bcl2-p53 binding. Blood, 2009. 113(2): p. 422-8. Deng, X., et al., Bcl2's flexible loop domain regulates p53 binding and survival. Mol Cell Biol, 2006. 26(12): p. 4421-34. Chipuk, J.E., et al., Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science, 2004. 303(5660): p. 1010-4. Cheng, J. and M. Haas, Frequent mutations in the p53 tumor suppressor gene in human leukemia T-cell lines. Mol Cell Biol, 1990. 10(10): p. 5502-9. Laumann, R., M. Jucker, and H. Tesch, Point mutations in the conserved regions of the p53 tumour suppressor gene not account for the transforming process in the Jurkat acute lymphoblastic leukemia T-cells. Leukemia, 1992. 6(3): p. 227-8. Pan, Y. and D.S. Haines, The pathway regulating MDM2 protein degradation can be altered in human leukemic cells. Cancer Res, 1999. 59(9): p. 2064-7. Knudson, C.M., et al., Bax-deficient mice with lymphoid hyperplasia and male germ cell death. Science, 1995. 270(5233): p. 96-9. Li, P.F., R. Dietz, and R. von Harsdorf, p53 regulates mitochondrial membrane potential through reactive oxygen species and induces cytochrome c-independent apoptosis blocked by Bcl-2. EMBO J, 1999. 18(21): p. 602736. Reinke, V. and G. Lozano, The p53 targets mdm2 and Fas are not required as mediators of apoptosis in vivo. Oncogene, 1997. 15(13): p. 1527-34. Chen, L., et al., Hydrogen peroxide-induced neuronal apoptosis is associated with inhibition of protein phosphatase 2A and 5, leading to activation of MAPK pathway. Int J Biochem Cell Biol, 2009. 41(6): p. 1284-95. Han, X., et al., Curcumin inhibits protein phosphatases 2A and 5, leading to activation of mitogen-activated protein kinases and death in tumor cells. Carcinogenesis, 2012. 33(4): p. 868-75. Zhang, Y., et al., Protein phosphatases 2A as well as reactive oxygen species involved in tributyltin-induced apoptosis in mouse livers. Environ Toxicol, 2012. Grundke-Iqbal, I., et al., Abnormal phosphorylation of the microtubuleassociated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A, 1986. 83(13): p. 4913-7. Oddo, S., et al., Abeta immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron, 2004. 43(3): p. 321-32. Murakami, K., et al., SOD1 (copper/zinc superoxide dismutase) deficiency drives amyloid beta protein oligomerization and memory loss in mouse model of Alzheimer disease. J Biol Chem, 2011. 286(52): p. 44557-68. 206 333. 334. 335. 336. 337. 338. 339. 340. 341. 342. 343. 344. 345. 346. 347. 348. 349. 350. Okamoto, K., et al., p53-dependent association between cyclin G and the B' subunit of protein phosphatase 2A. Mol Cell Biol, 1996. 16(11): p. 6593-602. Kong, M., et al., Alpha4 is an essential regulator of PP2A phosphatase activity. Mol Cell, 2009. 36(1): p. 51-60. Trockenbacher, A., et al., MID1, mutated in Opitz syndrome, encodes an ubiquitin ligase that targets phosphatase 2A for degradation. Nat Genet, 2001. 29(3): p. 287-94. Prickett, T.D. and D.L. Brautigan, Overlapping binding sites in protein phosphatase 2A for association with regulatory A and alpha-4 (mTap42) subunits. J Biol Chem, 2004. 279(37): p. 38912-20. Ruediger, R., H.T. Pham, and G. Walter, Alterations in protein phosphatase 2A subunit interaction in human carcinomas of the lung and colon with mutations in the A beta subunit gene. Oncogene, 2001. 20(15): p. 1892-9. Ruediger, R., H.T. Pham, and G. Walter, Disruption of protein phosphatase 2A subunit interaction in human cancers with mutations in the A alpha subunit gene. Oncogene, 2001. 20(1): p. 10-5. Tamaki, M., et al., PPP2R1B gene alterations inhibit interaction of PP2AAbeta and PP2A-C proteins in colorectal cancers. Oncol Rep, 2004. 11(3): p. 655-9. Soo Hoo, L., J.Y. Zhang, and E.K. Chan, Cloning and characterization of a novel 90 kDa 'companion' auto-antigen of p62 overexpressed in cancer. Oncogene, 2002. 21(32): p. 5006-15. Neviani, P., et al., The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein. Cancer Cell, 2005. 8(5): p. 355-68. Neviani, P., et al., FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia. J Clin Invest, 2007. 117(9): p. 2408-21. Dobrowsky, R.T., et al., Ceramide activates heterotrimeric protein phosphatase 2A. J Biol Chem, 1993. 268(21): p. 15523-30. Kim, S.W., et al., Ceramide produces apoptosis through induction of p27(kip1) by protein phosphatase 2A-dependent Akt dephosphorylation in PC3 prostate cancer cells. J Toxicol Environ Health A, 2010. 73(21-22): p. 1465-76. Kowluru, A. and S.A. Metz, Ceramide-activated protein phosphatase-2A activity in insulin-secreting cells. FEBS Lett, 1997. 418(1-2): p. 179-82. Nica, A.F., et al., Ceramide promotes apoptosis in chronic myelogenous leukemia-derived K562 cells by a mechanism involving caspase-8 and JNK. Cell Cycle, 2008. 7(21): p. 3362-70. Widau, R.C., et al., Protein phosphatase 2A (PP2A) holoenzymes regulate death-associated protein kinase (DAPK) in ceramide-induced anoikis. J Biol Chem, 2010. 285(18): p. 13827-38. Sawai, H., et al., Requirement of AP-1 for ceramide-induced apoptosis in human leukemia HL-60 cells. J Biol Chem, 1995. 270(45): p. 27326-31. Schutze, S., et al., TNF activates NF-kappa B by phosphatidylcholine-specific phospholipase C-induced "acidic" sphingomyelin breakdown. Cell, 1992. 71(5): p. 765-76. Conway, A., N.J. Pyne, and S. Pyne, Ceramide-dependent regulation of p42/p44 mitogen-activated protein kinase and c-Jun N-terminal-directed 207 351. 352. 353. 354. 355. 356. 357. 358. 359. 360. 361. 362. 363. 364. 365. 366. protein kinase in cultured airway smooth muscle cells. Cell Signal, 2000. 12(11-12): p. 737-43. Verheij, M., et al., Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis. Nature, 1996. 380(6569): p. 75-9. Aktas, O., et al., Fingolimod is a potential novel therapy for multiple sclerosis. Nat Rev Neurol, 2010. 6(7): p. 373-82. Cohen, J.A., et al., Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med, 2010. 362(5): p. 402-15. Khatri, B., et al., Comparison of fingolimod with interferon beta-1a in relapsing-remitting multiple sclerosis: a randomised extension of the TRANSFORMS study. Lancet Neurol, 2011. 10(6): p. 520-9. Liu, Q., et al., FTY720 demonstrates promising preclinical activity for chronic lymphocytic leukemia and lymphoblastic leukemia/lymphoma. Blood, 2008. 111(1): p. 275-84. Matsuoka, Y., et al., A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br J Pharmacol, 2003. 138(7): p. 130312. Chen, J., B.L. Martin, and D.L. Brautigan, Regulation of protein serinethreonine phosphatase type-2A by tyrosine phosphorylation. Science, 1992. 257(5074): p. 1261-4. Longin, S., et al., Selection of protein phosphatase 2A regulatory subunits is mediated by the C terminus of the catalytic Subunit. J Biol Chem, 2007. 282(37): p. 26971-80. Ogris, E., et al., A protein phosphatase methylesterase (PME-1) is one of several novel proteins stably associating with two inactive mutants of protein phosphatase 2A. J Biol Chem, 1999. 274(20): p. 14382-91. Ravizza, R., et al., The piperidine nitroxide Tempol potentiates the cytotoxic effects of temozolomide in human glioblastoma cells. Int J Oncol, 2004. 25(6): p. 1817-22. Darr, D., K.A. Zarilla, and I. Fridovich, A mimic of superoxide dismutase activity based upon desferrioxamine B and manganese(IV). Arch Biochem Biophys, 1987. 258(2): p. 351-5. Huber, K.R., et al., Superoxide dismutase-like activities of copper(II) complexes tested in serum. Biochim Biophys Acta, 1987. 915(2): p. 267-76. Lam, M.A., et al., Nitric oxide and nitroxides can act as efficient scavengers of protein-derived free radicals. Chem Res Toxicol, 2008. 21(11): p. 2111-9. Nagano, T., T. Hirano, and M. Hirobe, Superoxide dismutase mimics based on iron in vivo. J Biol Chem, 1989. 264(16): p. 9243-9. Weser, U., et al., Structure of Cu2(indomethacin)4 and the reaction with superoxide in aprotic systems. Biochim Biophys Acta, 1980. 631(2): p. 23245. Burger, J.A. and R.J. Ford, The microenvironment in mantle cell lymphoma: Cellular and molecular pathways and emerging targeted therapies. Seminars in Cancer Biology, 2011. 21(5): p. 308-312. 208 APPENDICES List of Publications Low, I.C., Z.X. Chen, and S. Pervaiz, Bcl-2 modulates resveratrol-induced ROS production by regulating mitochondrial respiration in tumor cells. Antioxid Redox Signal, 2010. 13(6): p. 807-19. Low, I.C., J. Kang, and S. Pervaiz, Bcl-2: a prime regulator of mitochondrial redox metabolism in cancer cells. Antioxid Redox Signal, 2011. 15(12): p. 2975-87. Krishna, S., I.C. Low, and S. Pervaiz, Regulation of mitochondrial metabolism: yet another facet in the biology of the oncoprotein Bcl-2. Biochem J, 2011. 435(3): p. 545-51. Coombs GS, Schmitt AA, Canning CA, Alok A, Low IC, Banerjee N, Kaur S, Utomo V, Jones CM, Pervaiz S, Toone EJ, Virshup DM , Modulation of Wnt/betacatenin signaling and proliferation by a ferrous iron chelator with therapeutic efficacy in genetically engineered mouse models of cancer. Oncogene, 2012. 31(2): p. 213-25. Conference Papers Low, I.C. and S. Pervaiz, Bcl-2 modulates resveratrol-induced ROS production by regulating mitochondrial respiration in tumor cells. European Molecular Biology Organization (EMBO) workshop on Mitochondria, Apoptosis & Cancer (MAC), Prague, Czech Republic. * Low, I.C. and S. Pervaiz, Bcl-2 as a regulator of mitochondrial respiration in tumor cells. International Cell Death Society annual meeting - South African chapter, Cape town, South Africa. ** Low, I.C. and S. Pervaiz, Nitration-mediated inhibition of Protein Phosphatase 2A as the mediator of superoxide-induced Bcl-2 phosphorylation. 18th Annual General Meeting of the Society for Free Radical Biology and Medicine (SFRBM), Atlanta, USA. *** * Recipient of Best Poster Award. ** Presented a short talk. *** Received travel award and was selected for a talk at the opening session. 209 [...]... Piperazine-N,N′-bis (2- ethanesulfonic acid) PKB/Akt Protein kinase B PKC Protein kinase C PMSF Phenylmethylsulfonyl fluoride PP1 Protein phosphatase 1 PP1-C Catalytic subunit of PP1 PP2A Protein phosphatase 2A PP2A- A A scaffolding subunit of PP2A PP2AB56α B56 -containing PP2A enzyme PP2AB56γ1 B56 1-containing PP2A holoenzyme PP2AB56δ B56 -containing PP2A PP2A-C Catalytic subunit of PP2A pRb Retinoblastoma protein Prx... xanthine and it is unlikely to be a major source of cellular H2O2 In fact, a major portion of cellular H2O2 is contributed by the dismutation of O2- into H2O2 by the superoxide dismutase (SOD) enzymes A total of three isoforms have been identified in eukaryotic cells to date, all of which are extremely efficient in their catalytic conversion of O2- into H2O2 Thus, with the constant production of O2-... contributing to the pool of cytosolic O2- in the cells 1.1 .2. 3 Xanthine oxidase and other potential sources of intracellular O2- Xanthine oxidase is an enzyme involved in the catabolism of purine where it catalyses the oxidation of hypoxanthine to xanthine, and subsequently to uric acid [35] During this process, O2 is being reduced into both O2- and H2O2 [1, 35] However, studies have shown that the majority... phosphorylation of Bcl- 2 did not affect Bcl- 2- Bak interaction 76 Figure 10: DDC-induced upregulation of S70 pBcl -2 is not a function of MAP kinases activation 79 Figure 11: PP2A activity declined upon pharmacological inhibition or genetic silencing of SOD1 81 Figure 12: DDC inhibited the interaction between PP2A- C and Bcl- 2 84 Figure 13: DDC inhibited the co-localization of PP2A- C... arsenal of antioxidant machineries capable of eradicating H2O2 2. 2 Catalase Catalase is a haem-protein responsible for the neutralization of intracellular H2O2 [87, 88] It is widely expressed in most tissues, particularly in the liver Like SOD, catalase catalyses a dismutation reaction, whereby one H2O2 molecule is reduced to H2O while another H2O2 molecule is simultaneously oxidized to form O2 in a single... B56 and Bcl- 2 was not affected by DDC treatment 99 xii Figure 19: Silencing of SOD1 inhibited PP2A- C -B56 interaction, but not the interaction between Bcl- 2 and B56 101 Figure 20 : SiRNA -mediated knockdown of SOD1 inhibited mitochondrial translocation of PP2A- A and PP2A- C, but not B56 103 Figure 21 : DDC treatment resulted in a decline in intracellular NO level 105 Figure 22 : FeTPPS... O2- from the mitochondria as well as other enzymatic sources, it is of no surprise that the majority of cellular H2O2 stems from the dismutation of these O2- by the SODs Due to their role in the removal of O2-, SODs are considered as part of the cellular antioxidant machinery, and for this reason, a more detailed discussion of the enzyme in the following chapter on cellular antioxidant systems is warranted... it [1] 1 .2. 2 Sources of intracellular H2O2 Unlike the production of O2- by the NOX complexes, no enzyme is known to intentionally produce H2O2 in the cell In vivo, H2O2 are typically produced as byproducts of the various biological processes catalysed by the oxidase enzymes For instance, the mitochondrial monoamine oxidases are amongst the major contributors of mitochondrial H2O2 The monoamine oxidases... and Bcl- 2 87 Figure 14: SiRNA -mediated silencing of SOD1 inhibited the co-localization of PP2A- C and Bcl- 2 89 Figure 15: DDC treatment inhibited mitochondrial translocation of PP2A- C 93 Figure 16: HA -B56 interacts with Bcl- 2 96 Figure 17: DDC treatment inhibited the interaction of PP2A- C with the B56 regulatory subunit 98 Figure 18: Co-localization of B56 ... DDC-induced upregulation of S70 pBcl -2 106 Figure 23 : Low doses of ONOO- induced the phosphorylation of Bcl- 2 at S70 108 Figure 24 : Low doses of ONOO- protected against drug-induced cell death 109 Figure 25 : ONOO- treatment inhibited the interaction of Bcl- 2 with the AC catalytic core but not with the B56 regulatory subunit 111 Figure 26 : Pre-treatment of FeTPPS blocked the . subunit of PP1 PP2A Protein phosphatase 2A PP2A- A A scaffolding subunit of PP2A PP2A B56 B56 -containing PP2A enzyme PP2A B56 1 B56 1-containing PP2A holoenzyme PP2A B56 B56 -containing. level of S70 pBcl -2 by inhibiting the holoenzyme assembly of B56 -containing PP2A (PP2A B56 ) 94 3.1 The B56 regulatory subunit of PP2A is responsible for the substrate recognition of Bcl- 2 in. knockdown of SOD1 triggered a drop in PP2A activity 80 2. 3 O 2 - inhibited the interaction between Bcl- 2 and the catalytic subunit of PP2A 82 2. 4 Co-localization of PP2A- C and Bcl- 2 was inhibited

Ngày đăng: 09/09/2015, 10:14

Mục lục

  • HT081818N Ivan Low Cherh Chiet Amended PhD Thesis 2012

Tài liệu cùng người dùng

Tài liệu liên quan